Skip to main content

Full text of "Specific-mutational patterns of p53 gene in bladder transitional cell carcinoma among a group of Iraqi patients exposed to war environmental hazards."

See other formats


Al-Kashwan et al. BMC Research Notes 2012, 5:466 
http://www.biomedcentral.eom/1756-0500/5/466 



Research Notes 



RESEARCH ARTICLE Open Access 



Specific-mutational patterns of p53 gene in 
bladder transitional cell carcinoma among a 
group of Iraqi patients exposed to war 
environmental hazards 

Thekra A Al-Kashwan 1 , Massoud Houshmand 2 , Asaad Al-Janabi 3 , Alice K Melconian 4 , Dhafir Al-Abbasi 3 , 
Muhammad N Al-Musawi 5 , Maryam Rostami 2 and Akeel A Yasseen 3,6 * 



Abstract 

Background: To unfold specific-mutational patterns in TP53 gene due to exposures to war environmental hazards 
and to detect the association of TP53 gene alteration with the depth of bladder cancer. 

Methods: Twenty-nine bladder carcinomas were analyzed for TP53 alterations. PCR-single strand conformational 
polymorphism analysis, DNA sequencing and immunohistochemical analysis using monoclonal mouse anti-human 
p53 antibody (Clone DO-7) were employed. 

Results: TP53 gene mutations occurred in 37.9% of the cases while TP53 overexpression occurred in 58.6%. Both of 
them were associated with deep invasive-tumors. Single mutations were seen in 63.6%, whereas only 27.3% have 
shown double mutations. Four mutations were frameshifted (30.8%); two of them showed insertion A after codon 
244. There was no significant association between TP53 mutations and protein overexpression (P>0.05), while a 
significant association was observed between TP53 alterations and tumors progression (P < 0.01). 

Conclusion: The infrequent TP53mutations, especially insertion A and 196 hotspot codon, may represent the 
specific-mutational patterns in bladder carcinoma among the Iraqi patients who were exposed to war 
environmental hazards. TP53 alteration associated with bladder cancer progression should be analyzed by both 
mutational and protein expression analysis. 

Keywords: Bladder cancer, TP53 alteration, Specific mutation, Immunohistochemistry 



Background 

Bladder cancer (BC) is the most common malignancy 
affecting urinary system comprising the seventh most 
common cancer worldwide with male predominance 
[1,2]. In Iraq, the incidence of bladder cancer is the 
fourth most common type of cancer (in men and eighth 
most common in women) [3]. The most common type 
of bladder cancer is transitional cell carcinoma (TCC) 
accounting for more than 90% of the cases [2]. It repre- 
sents one of the first tumors that have been associated 



* Correspondence: a.yasseen@hotmail.com 

department of Pathology and Forensic Medicine, Faculty of Medicine, 
University of Kufa, Kufa, Iraq 

department of Pathology and Forensic Medicine, Faculty of Medicine, 

University of Kufa, Kufa, P.O. Box 21, Najaf Governorate, Iraq 

Full list of author information is available at the end of the article 

(3 BioMed Central 



with environmental risk factors that produce genetic 
alterations [4,5]. 

TP53 gene "the guardian of genome" is the most fre- 
quently mutated tumor suppressor gene identified in 
human cancer. TP53 inactivation led to diminished con- 
trol cell cycle check points, decreased DNA repair, and 
increased genomic instability [6-8] . Furthermore, TP53 
inactivation has been identified in tumor progression, 
metastasis, and aggressive phenotype of bladder cancer 
giving rise to what has been considered as a useful gen- 
etic biomarker to predict progression associated with 
bad prognosis [9-11]. The frequency and the type of 
mutations vary from one tumor type to another, ranging 
from 5% to 80% depending on the type, stage and eti- 
ology of tumor [12]. Almost all TP53 mutations are 



© 2012 Al-Kashwan et al.; licensee BioMed Central Ltd. This is an Open Access article distributed under the terms of the 
Creative Commons Attribution License (http://creativecommons.Org/licenses/by/2.0), which permits unrestricted use, 
distribution, and reproduction in any medium, provided the original work is properly cited. 



Al-Kashwan et al. BMC Research Notes 2012, 5:466 
http://www.biomedcentral.eom/1756-0500/5/466 



Page 2 of 10 



point missense mutations leading to a functionally de- 
fective protein [13]. Approximately 90% of them are 
localized in DNA-binding domains encoded by exons 5- 
8. In total, about 40% of them are localized at the "hot- 
spot" residues R 175, G 245, R 248, R 249, R 273 and R 
282 [14]. Indeed, there are considerable numbers of 
studies on TP53 gene being as a target for carcinogens, 
with a specific TP53 mutations spectrum, mainly G: 
C^A: T transitions at CpG and non-CpG sites in blad- 
der cancer [7,15,16]. It is well documented that the 
spectrum of TP53mutations in bladder cancer differs 
from that of lung cancer, even though cigarette smoking 
is probably a contributing cause in over one-third of all 
bladder cancer cases [17]. Specifically, G^T transver- 
sions at CpG are relatively uncommon (about 8% in 
bladder cancer versus 27% in lung cancer) whereas CpG 
G: C^A: T transition are as twice as common with re- 
gard to mutation patterns (22% for bladder cancer ver- 
sus 11% for lung cancer) [7]. 

Inactivation of tumor suppresser genes can occur ei- 
ther primarily through mutations, or without any change 
in the structure of the given genes. Thus, tumor sup- 
pressor function should be analyzed at the level of the 
genes as well as at the level of proteins, and in the con- 
text of the pathways in which these genes are involved 
[18,19]. The mutated p53 protein has a longer half-life, 
as compared with normal p53 protein, which can be 
detected by Immunohistochemistry (IHC) as a surrogate 
marker for mutation [20]. Immunohistochemical positiv- 
ity for TP53protein is in general thought to reflect point 
mutations of TP53 gene in tumor, although it is not al- 
ways synonymous with TP53 mutation [21]. Accordingly, 
using both molecular and protein analyses (PCR-based 
genetic technique and immunohistochemistry respect- 
ively) for detection of TP53 alterations have rational effi- 
cacy for mutation detection rather than each one alone 
[20,22,23]. The aims of this research were to determine 
if any specific-mutational pattern in TP53gene may play 
a possible role in bladder tumorogenesis, which might 
be resulted from the exposure to the hazardous pollu- 
tion of the wars in Iraq. An analysis of the alteration of 
TP53gene using combined PCR-based genetic and IHC 
analysis to evaluate its association with bladder cancer 
progression was conducted. 

Methods 

Ethical approval was obtained from the local medical 
ethics committee, Faculty of Medicine, University of 
Kufa. A written informed consent was received from all 
subjects before proceeding any further. The study was 
designed and conducted in accordance with the tents of 
Declaration of Helsinki. 

A total of twenty-nine patients (25 males and 4 females) 
with transitional cell carcinoma (TCC), diagnosed by 



transurethral resection (TUR-biopsy) at the Department 
of Pathology of Kufa School of Medicine Teaching Hos- 
pital were subjected to the present study. The patients 
were randomly selected from the Middle Euphrates area 
and south of Iraq. Both regions were potentially exposed 
to environmental pollution during the last two decades of 
wars. The patients ages ranged between 35 and 85 years, 
with a median age of 69.3 years. Histological examination, 
grading [24] and staging [25] were performed by two of us 
independently. Three were classified as grade I, two as 
grade II, and fifteen classified as grade III. Fourteen cases 
were superficial TCC [5 as Ta and 9 as Tl], while 15 cases 
were deep invasive TCC (T2). Sixteen of our patients had 
a history of cigarette smoking for at least 10 years. 

TP53 status 

TP53 protein expression 

TP53 protein expression was estimated using immuno- 
histochemical techniques [26-28]. Five micron thick sec- 
tions of formalin-fixed paraffin-embedded tissue (FFPE) 
were placed on positively charged slides (Fisher scientific 
Co., Pittsburgh, PA). These sections were then deparaffi- 
nized and rehydrated. For staining enhancement, the 
sections were pre-treated with antigen retrieval solution 
(0.01 M, citrate buffer, pH6.0, Dako Cytomation/Den- 
mark) in water-bath at 95°C for 40 minutes followed by 
staining with a monoclonal mouse anti-human TP53 
antibody (Clone DO-7, Ready-to-use, DakoCytomation/ 
Denmark). The antigen-antibody complex was visualized 
using Labeled Streptavidin-Biotin 2 System-Horseradish 
Peroxidase staining technique (LAB/LSAB2 System- 
HRP, DakoCytomation/Denmark). The sections were 
then counterstained with Meyer's haematoxylin. 

A breast cancer with high TP53expression detected by 
immunohistochemical analysis was used as positive ex- 
ternal control. Negative controls were obtained by omis- 
sion of the primary antibody and by a breast cancer with 
negative TP53 expression detected by immunohisto- 
chemical analysis. The non-epithelial cells of samples 
(lymphocytes, stromal cells and endothelial cells) were 
used as negative internal control. 

All slides were reviewed independently by two investi- 
gators without previous knowledge of tumor grade and 
stage or TP53mutation. TP53 expression (nuclear stain- 
ing) was evaluated by counting 100 cells/section in five 
randomly chosen high-power fields (40x) by light micro- 
scope. The extent of nuclear reactivity was classified in 
four categories [9,20]; no nuclear reactivity (-), few fo- 
cally positive cells (1 to 10% tumor cells) (+/-), hetero- 
geneous nuclear reactivity (10 to 50% tumor cells) (+) 
and homogenous intense nuclear reactivity (50 to 100% 
tumor cells) (++). The samples which demonstrated at 
least 10% nuclear reactivity were considered to be TP53- 
positive (have an alteration in TP53) [9,10,19,20]. 



Al-Kashwan et al. BMC Research Notes 2012, 5:466 
http://www.biomedcentral.eom/1756-0500/5/466 



Page 3 of 10 



TP53 mutation 
DNA extraction 

High-molecular weight DNA was prepared from the 
fresh tumor specimens by phenol-chloroform-isoamyl 
alcohol method [20,29]. DNA which was extracted from 
whole blood samples of healthy looking individuals was 
included as normal control with their matched age and 
sex with Wizard ® Genomic DNA purification kit (Pro- 
mega Company/USA). 

PCR-SSCP and DNA sequencing 

The mutational analysis was achieved using PCR-single 
strand conformation polymorphism (PCR-SSCP) and 
direct DNA sequencing methods for TP53 gene exons 
5-8. Each exon 5-8 was amplified by PCR. The primer 
sequences used were as follows [15]: (1) exon 5/228 bp, 
forward: 5/-TTCAACTCTGTCTCCTTCCT-3/ and re- 
verse: 5/-CAGCCCTGTCGTCTCTCCAG-3/; (2) exon 
6/159 bp, forward: 5/- GCCTCTGATTCCTCATCGAT- 
3/ and reverse:5/-TTAACCCCTCCTCCCAGAGA-3/; 
(3) exon 7/157 bp, forwarder- AGGCGCACTGGCCT- 
CATCTT-3/ and reverse:5/-TGTGCAGGG TGG 
CAAGTGGC-3/; and (4) exon 8/214 bp, forward:5/- 
TTCCTTACTGCCTCTTGCTT-3/and reverse: 5/- 
AGGCATAACTGCAC CCTTGG-3/. Each PCR reaction 
was performed in a final volume of 25 ul containing 
100 ng DNA, IX PCR buffer, 1.5 mM MgCL2, 200 uM 
each dNTP, 0.1 uM of each upstream and downstream 
primer, and 1.5u of Taq polymerase (CinnaGen Com- 
pany/Iran). PCR was carried out under the following 
conditions: an initial denaturation step (95°C for 5 min- 
utes) was followed by 35 cycles consisting of (for exon 5) 
denaturation at 95°C for 50 seconds, primer annealing at 
55°C for 35 seconds and extension at 72°C for 30 sec- 
onds; (for exons 6-7) denaturation at 95°C for 50 sec- 
onds, primer annealing at 63°C for 25 seconds and 
extension at 72°C for 15 seconds; or was followed by 
30 cycles (for exons 8) consisting of denaturation at 95° 
C for 50 seconds, primer annealing at 65°C for 25 sec- 
onds and extension at 72°C for 15 seconds. The final ex- 
tension was continued for 10 minutes at 72°C. The PCR 
products were analyzed on 1.5-2% agarose gel to deter- 
mine the specific band of each exon product and then 
analyzed on 12% polyacrylamide gel to another evalu- 
ation of specific band purity to ensure the absence of 
any unwanted products (non-specific bands) which may 
interfere with SSCP and DNA sequencing analysis. 

For non-radioactive SSCP analysis, the SSCP analysis 
was carried out according to the method of Liechti-Gallati 
et al, (1999) [30], for both of the tumor and the control 
samples. 

The to controls were analyzed. PCR products were 
denaturated at 96°C for 10 min at 3:2 dilution of forma- 
mide loading dye (SSCP denaturing solution) containing 



95% formamide, 100 mM NaOH, 0.25% bromophenol 
blue, 0.25% xylene cyanol and thereafter placed immedi- 
ately on ice to prevent re-annealing of the single- 
stranded product. The denaturized samples, the controls 
and the normal PCR product of controls were loaded 
quickly into wells of the 12% polyacrylamide gel and run 
at 4-10°C/80 V for overnight. The gel was stained with 
Sliver Staining and alterations of bands relative 

All the samples that revealed mobility shift in their mi- 
gration during SSCP screening mutation analysis were 
sent to the direct DNA sequencing in both directions: 
forward sequencing 5/^3/and reverse sequencing 
3/^5/using the same primer sequences and dideoxy 
chain termination method. The DNA sequencing was 
achieved by Gen Fanavaran Company/Iran. The DNA 
sequencing chromatogram was interpreted using Chro- 
magen 2.3 version software that allows comparison of a 
newly generated sequence of DNA sample with free 
DNA sequence. Viewers (available from National Center 
for Biotechnology Information (NCBI) website) were 
used as reference sequence of gene of interest for 
comparison. 

Statistical analysis 

The Fishers exact probability test and Odds ratios 
(ORs), using contingency tables, were applied to analyze 
the data using the program statistical package for the 
Social Science (SPSS for windows, version 10.0). The 
relationships between the variables were assessed using 
non-parametric Fisher s exact probability test. A P-value 
<0.05 was considered as statistically significant at a level 
of 5%. The strength of the associations between the vari- 
ables was measured by calculating Odds ratios (ORs) 
and confidence intervals (95% CI). Possible categories 
for OR are greater than land less than 1. A value greater 
than 1 indicates positive association and a value less 
than 1 indicates negative association. 

Results 

Mutational analysis of the TP53 gene in relation to 
clinicopathological features 
Characteristic of TP53 gene mutation 

The PCR-SSCP analysis showed that 12 out of 29 cases 
of bladder cancer patients had aberrantly migrating 
bands or extra bands that were further analyzed by DNA 
sequencing to represent the mutations of TP53 gene 
(Figure 1). All controls used in this study revealed nor- 
mal migration of SSCP bands and normal sequencing of 
all studied exons (Figure la,c,e). 

The sequencing results confirmed that 10 cases har- 
bored one or more TP53 mutations within identical or 
separated samples, one case had silent mutation and one 
case had bad sequencing. Eleven (37.9%) cases were clas- 
sified as TP53-positive [10 cases with detected mutation 



Al-Kashwan et al. BMC Research Notes 2012, 5:466 
http://www.biomedcentral.eom/1756-0500/5/466 



Page 4 of 10 





Figure 1 Example of PCR-SSCP/HD and DNA sequencing analysis of p53 gene in transitional cell carcinoma, (a) PCR-SSCP/HD analysis of 
exon 5 shows extra bands and bands with mobility shift indicated by arrows (-ve: undenaturated DNA control; +ve: denaturated DNA control, 
and T: tumors). DNA sequencing reveals transition CGC^CAC at codon 175 in exon 5 (b) and insertion A after codon 244 (GGC) in exon in exon 
7 (d) in compared to wild types (c) and (e) respectively (arrows). 



and 1 case without DNA sequencing] and 18 (62.1%) 
cases as /?53-negative [17 cases without detected muta- 
tion and 1 case with silent mutation: case 5, codon 244; 
GGC (Gly) to GGA (Gly)] (Table 1). 

Among TP53-positive cases; seven patients (63.6%) 
showed single mutation, three patients (27.3%) had 
double mutations and one patient (9.1%) with mutation 
detected only by SSCP analysis. A total of 13 mutations 
determined in ten cases; 9 mutations (69.2%) were 



single-base pair substitutions and 4 mutations (30.8%) 
were with frameshift mutations. 

The patterns of TP53 base-pair mutations showed that 
five mutations (38.4%) were of transitions including G: C 
— »A: T, three of them (23.8%) occurred at CpG di- 
nucleotide in the codons (175 and 196) that were 
reported as hotspot for TP53 mutations. The other two 
(15.4%) mutations occurred at non-CpG sites. The four 
(30.8%) transversions detected were two (15.4%) G — > C, 



Al-Kashwan et al. BMC Research Notes 2012, 5:466 
http://www.biomedcentral.eom/1756-0500/5/466 



Page 5 of 10 



Table 1 Mutational analysis of the p53 detection by SSCP and DNA sequencing, p53 nuclear reactivity and p53 status 



Patients No Exon/Codon Codon change Base change Mutation type Amino acid change p53 nuclear reactivity p53 status 



1 

2 


- 


- 


- 




- 


- 


- 


3 


5/184 


GAT^AAT 


G^A 


Ts 


Asp184Asn 


++ 


+ 


4 


- 


- 


- 




- 


- 


- 


5 


7/244 


GGC^GGA 


C^A 


Tv 


Gly244Gly 


- 


- 


6 


7/230 


Del A 


- 


Fr 


- 


- 


+ 


7 
8 


7/NS 


- 


- 




- 


++ 


+ 


9 


- 
- 


- 
- 


- 
- 




- 
- 


- 

++ 


- 

+ 


10 


- 


- 


- 




- 


++ 


+ 


11 


- 


- 


- 




- 


- 


- 


12* 


5/154 


GGC^GGA 


C^A 


Tv 


Gly154Gly 


+ 


+ 




6/192 


CAG^CAC 


G^C 


Tv 


Glu192His 






13 


7/244 


+ A 


- 


Fr 


- 


- 


+ 


14 


- 


- 


- 


- 


- 


- 


- 


15 


7/244 


+ A 


- 


Fr 


- 


- 


+ 


16 


- 


- 


- 


- 


- 


++ 


+ 


17 


- 


- 


- 


- 


- 


+ 


+ 


18 


- 


- 


- 


- 


- 


+ 


+ 


19 


- 


- 


- 


- 


- 


++ 


+ 


20* 


6/196 


CGA^CAA 


G^A 


Ts 


Arg196Glu 


++ 


+ 




8/283 


CGC^CCC 


G^C 


Tv 


Arg283Pro 






21 


5/175 


CGC^CAC 


G^A 


Ts 


Arg175His 


++ 


+ 


22 


5/176 


TGC^GGC 


T^G 


Tv 


Cys176Gly 


++ 


+ 


23 












++ 


+ 


24 
















25* 


8/293 


Del G 




Fr 






+ 




8/294 


GAG^GAA 


G^A 


Ts 


Glu294Glu 






26 


6/196 


CGA^CAA 


G^A 


Ts 


Arg196Glu 


++ 


+ 


27 












++ 


+ 


28 












++ 


+ 


29 












++ 


+ 



p53 status; + has alteration (mutation) or immunoreactivty +,++ (IHC), -; no alteration (mutation (-) and IHC (-,+/-), Ts; Transition, Tv; Transvertion, *; Tumor with 
double mutation, NS; DNA not sequenced but p53 mutation detected by SSCP analysis, - G; deletion of one base (G) at codon 293, - A; deletion of one base (A) at 
codon 230, + A; Insertion of one base (A) after codon 244/exon 7, Fr; Frameshift. 



one (77%) T^G, and one (7.7%) C^A. Two of the 
base-pair substitutions were silent mutations found in 
double mutations [case 12, codon 154; GGC (Gly) to 
GGA (Gly) and case 25, codon 294; GAG (Glu) to GAA 
(Glu)], and seven were missense resulted in amino acid 
changes. 

Of the observed frameshift mutations (30.8%) three 
(23.1%) were in the exon7 [two (15.4%) with insertion A 
after the codon 244 (GGC/Gly) and one (7.7%) with de- 
letion A at the codon 230(ACC/Thr)]. The remaining 



one was deletion G (7.7%) at codon 293 (GGG/Gly) in 
exon 8. The double mutations were found in case 12 in 
exon 5 [codon 154; Gly — ► Gly (silent)] and in exon 6 
[codon 192; Glu —> His (missense)], case 20 in exon 6 
[codon 196; Arg^Glu (missense)] and exon 8 [codon 
283; Arg^pro (missense)], and case 25 in exon 8 
[codon 293, del G and codon 294, Glu -> Glu (silent)] 
(Table 1) (Figure lb and d). The TP53 mutations were 
higher in deep invasive-tumors (high grade and stage 
T2) (40%) than in superficially invasive tumors (low 



Al-Kashwan et al. BMC Research Notes 2012, 5:466 
http://www.biomedcentral.eom/1756-0500/5/466 



Page 6 of 10 



grade and stage Ta-Tl) (35.7% 
(Table 2). 



(OR, 1.2; CI, 0.26-5.4) 



Immunhistochemical analysis of TP53 gene in relation to 
clinicopathological features 

Immunohistochemical analysis showed that TP53 immu- 
noreactivty was restricted to the nuclei of tumor cells. 
Four patterns of immunohistochemical nuclear staining 
was observed (Figure 2) including: no detectable immu- 
noreactivty (-) in 20.7% (Figure 2,a), few focal reactivity 
in less than 10% of tumor cells (+/-) in 20.7% (Figure 2, 
b), heterogeneous nuclear reactivity in 10 -50% of tumor 
cells (+) in 10.3% (Figure 2,c), and intense homogenous 
nuclear reactivity was greater than 50% of tumor cells (+ 
+) in 48.3% (Figure 2,d). The level of TP53 nuclear re- 
activity was classified into two categories: wild- type 
TP53including score (-) and (+/-), and altered TP53 
includes score (+) and (++). While, negative internal 
control of samples (non-epithelial cells) showed absence 
of TP53 nuclear staining that was consistent with wild- 
type expression of TP53 gene (Figure 2, D). The TP53 
overexpression was identified in 17 cases (58.6%) out of 
29 TCC cases. The altered expression of TP53 was more 
frequent in high grade tumor than low grade (66.7% vs. 
28.6% respectively) and in T2 stage than Ta and Tl 
(66.7% vs. 28.6% respectively), giving rise to a statistically 
significant difference (p < 0.05) (Table 2). 

Correlation of p53 mutation with nuclear reactivity 

The present data shows no significant association be- 
tween TP53 mutations and the immunohistochemical 
detection of TP53 protein (p>0.05) (Table 3). Seven 
tumors which had TP53 mutation [six of them with 
point mutation (missense), and one case with SSCP mo- 
bility shift] were positive for TP53protein overexpres- 
sion. Four tumor cases with frameshift mutations of 
TP53had no detectable TP53 nuclear accumulation. Ten 



tumor cases without detected mutation had TP53over- 
expression. Accordingly, the TP53 alteration (TP53 sta- 
tus) defined by either presence of mutations or TP53 
immunoreactivty, or both was demonstrated (Table 1) 
[23]. Our data showed that the TP53 alteration (TP53 
status) was strongly associated with high grade and stage 
(p< 0.01) (Table 2). 



Discussion 

In Iraq, the incidence of most types of cancer (including 
bladder cancer) has increased sharply in the last few 
years due to exposure to wars pollution [31-33]. Muta- 
tional analysis of the TP53 gene provides a unique op- 
portunity to investigate the etiology, epidemiology, and 
pathogenesis of human cancer [34,35]. Few studies have 
been conducted on the Iraqi population following the 
conflicts that led to hazardous environmental pollution. 
The effects of exposure to such pollutants is hard to as- 
certain as it is not possible to compare to data pre-con- 
flict, which are not available either because they were 
not measured or, because they were destroyed. There is 
also the difficulty in finding a cohort of unexposed indi- 
viduals: the conflicts were country wide and the shifting 
sands spread chemical pollutants over great distances. A 
definitive geographical location of where the greatest 
chemical pollution exists could not be determined 
[31,32], However, it is not implausible to suggest that al- 
most all Iraqis have had some exposure to hazardous 
and/or toxic substances. To the best of our knowledge, 
the present study is the first molecular analysis of TCC 
in Iraq to determine the pattern of TP53 mutations. The 
results were compared with other areas polluted with 
radioactive substances [15,35] and with reported specific 
TP53 mutations that were known to be associated with 
smoking, as smoking is a major risk factors for bladder 
cancer [5]. 



Table 2 Association between p53 mutations as analyzed by SSCP and DNA sequencing, p53 immunoreactivty and p53 
status with clinicopathological features 



Variable Patients No. p53 mutation (5-8 exons) No. (%) 



p53 nuclear reactivity No. (%) 



p53 mut p53 wt OR 95%CI 



Negative 



Positive 



p53 status No. (%) 

P value altered Non- P value 
altered 



+/- 



Total 



Total 



Total 


29 


1 1 (37.9) 


18(62.1) 


6(20.7) 


6(20.7) 


12(41.4) 


3(10.3) 


14(48.3) 


1 7(58.6) 


21(72.4) 


8(27.6) 


Grade 
























Low(l&[[) 


14 


5(35.7) 


9(64.3) 


4(28.6) 


5(35.7) 


9(64.3) 


1(7.1) 


4(28.6) 


5(35.7) 


0.02 7(50) 


7(50) 










1 .2 0.26-5.4 














0.01 


High (III) 


15 


6(40) 


9(60) 


2(13.3) 


1(6.7) 


3(20) 


2(13.3) 


10(66.7) 


12(80) 


14(93.3) 


1(6.9) 


Stage 
























LowO"a&T1) 


14 


5(35.7) 


9(64.3) 


4(28.6) 


5(35.7) 


9(64.3) 


1(7.1) 


4(28.6) 


5(35.7) 


0.02 7(50) 


7(50) 










1 .2 0.26-5.4 














0.01 


High (T2) 


15 


6(40) 


9(60) 


2(13.3) 


1(6.7) 


3(20) 


2(13.3) 


10(66.7) 


12(80) 


14(93.3) 


1(6.9) 



Negative indicates that less than 10% of tumor nuclei demonstrated p53 immunoreactivty, positive; 10% or more of tumor nuclei demonstrated p53 
immunoreactivty, p53 status; + has alteration (mutation, or immunoreactivty score +,++, or both), - no alteration (mutation (-) and IHC (score -, +/-) for p53), OR, 
Odds Ratio; CI, Confidence Intervals, P Value < 0.05 or 0.01 (Fisher's exact probability test). 



Al-Kashwan et al. BMC Research Notes 2012, 5:466 
http://www.biomedcentral.eom/1756-0500/5/466 



Page 7 of 10 




Figure 2 Immunohistochemical detection of p53 nuclear reactivity in transitional cell carcinoma (a) Invasive transitional cell carcinoma, 
poorly differentiated (Grade III) showing no detectible nuclear p53 immunostaining (Score (-), (10X)). (b) Papillary transitional cell 
carcinoma, well differentiated (Grade I) showing a few p53 immunoreactivty of tumor nuclei (Score (+/-), arrowed, (10X and 40X)). (c) Invasive 
transitional cell carcinoma, poorly differentiated (Grade II) showing heterogeneous p53 nuclear immunostaining (Score (+); arrowed,(40X)). (d) 
Papillary transitional cell carcinoma, well differentiated (Grade I) showing homogenous intense p53 nuclear immunostaining (Score (++); yellow 
arrow, (10X and 40X), (Red arrow indicates surrounding stromal and infiltrative lymphocytes with no detectible nuclear p53 immunostaining)). 



The present investigation revealed that the frequency 
of TP53 mutations was higher in deep invasive-tumors 
(high grade and stage T2) than superficially invasive 
tumors (low grade and stage Ta-Tl) (37.9% Vs 35.7%) 
(OR, 1.2; CI, 0.26-5.4) (Table 2). These observations pro- 
vide further support for the proposed association be- 
tween TP53 alteration and bladder cancer progression 
[5,20,36]. In these results, a history of smoking was not 
associated with a high frequency of TP53 mutation, nor 
with the pattern of mutation in patients with TCC. 



Table 3 Relation of p53 immunoreactivty to p53 



mutation analyzed by SSCP and DNA sequencing 





Total 


p53 nuclear reactivity No. (%) 
Positive Negative 


P 

value 


p53 mutation No. (°/ 


3) 29 


1 7(58.6) 


12(41.4) 




p53 mut 


1 1 (37.9) 


7(63.7) 


4(36.3) 


0.09 


p53 wt 


18(62.1) 


10(55.6) 


8(44.4) 





P-value> 0.05 (Fisher's exact probability test); Mut, mutant; wt, wild-type. 



Although the number of our patients is rather small, 
many interesting observations are apparent. Our results 
showed that the TP53 double mutations constituted 
27.3% of all mutations detected, which is consistent with 
values reported by Yamamoto et al, (1999) [15] who 
found double mutations in their cases of dysplasia and 
CIS in radio-contaminated areas. The possible explan- 
ation for the occurrence of double mutations found in 
our study is most likely because of the presence of a 
strong carcinogenic insult from war pollution that may 
have resulted in multiple transformation events [15,37]. 

It has been indicated that G — > A transitions were the 
most prevalent type of TP53 mutation in bladder cancer; 
about half of these transitions occurred at CpG sites 
[38]. There is increasing evidence to indicate that CpG 
bases might be more susceptible than other sites to at- 
tack by environmental mutagens [39] . Studies conducted 
in radio-contaminated regions in Ukraine found a high 
frequency of CpG/G — > A mutations (73%) that were sig- 
nificantly different from those reported by I ARC [17]. In 
the present study, the frequency of all G —> A transitions 



Al-Kashwan et al. BMC Research Notes 2012, 5:466 
http://www.biomedcentral.eom/1756-0500/5/466 



Page 8 of 10 



was 38.4% and those that occurred at CpG sites were 
23.8% with predominant hotspots (2 out of 3 cases) at 
codon 196. This is considered to be infrequent in blad- 
der tumors [40] and may reflect the effect of certain ex- 
ogenous carcinogens, such as depleted uranium, on the 
frequency of mutation (Table 1). 

Conversely, the transversion type of mutation that was 
shown in this study (30.8%) was similar to a previously 
reported finding which confirmed that TP53 mutation in 
bladder cancer patients who smoked consisted of G:C —> 
C:G transversions [41,42]. In our study, most TP53 
mutations occurred in both smokers and non-smokers 
(such as G — ► C transversions), or occurred in non- 
smokers only (such as C —> A and T — > G transversions). 
This observation unveils new evidence for the presence 
of risk factors for cancer, other than cigarette smoking, 
that may underlie this type of mutation. 

Another important observation was the frameshift 
mutations which were found to be higher (30.8%) in fre- 
quency than what has been reported previously [5,43], 
and was found preferentially to occur in exon-7 . Two 
the frameshift mutation having the same mutation (in- 
sertion A after codon 244) that cannot be excluded as a 
relative hotspot (Table 1). In fact, the most common 
mutations observed in TP53 DNA binding (exons 5-8) 
were missense mutations while frameshift types were 
found to be less frequent [43]. Most studies concerning 
the mutational signature of p53 in relation to the history 
of smoking revealed that the frameshift mutation in the 
DNA binding either have not been observed [44] or oc- 
curred less frequently than point mutation (7% and 
7.1%) with different types of deletions and insertions 
[7,40]. In radio-contaminated regions in Ukraine, all 
TP53 mutations determined were single-bp substitu- 
tions; no base deletions or insertions were found [15,45]. 
This indicated a possible distinct molecular carcinogen- 
esis pathway for bladder cancer after the Chernobyl dis- 
aster, based on a different incidence of p53 gene 
mutations compared with tumors found in the same 
population before the accident [45]. 

Accordingly, frameshift mutations (especially insertion 
A) of TP53 may reflect the effect of certain exogenous 
environmental contamination and may be considered as 
a useful predictor marker for bladder cancer. No reports 
on the levels of environmental pollution from toxic che- 
micals or radiation exist as no studies have been con- 
ducted to measure and specify these effects with 
accuracy. This study and future studies on the health of 
the Iraqi populace are needed to stimulate investigations 
on the extent and localization of environmental pollu- 
tion caused by the recent conflicts. 

In fact, the occurrence of TP53mutations leads to con- 
formational changes of the protein, resulting in a pro- 
longed half-life and subsequent accumulation of mutations 



in the nuclei. The extended half-life of the protein is the 
basis for immunohistochemical detection of TP53 
[20,22,44]. Whilst immunohistochemical positivity for 
TP53 protein generally reflects point mutations of TP53 
genes in tumor cells, it is not always synonymous with 
mutations [20,22]. The present study revealed that the 
TP53 overexpression (58.6%) was observed more fre- 
quently in high grade and in T2 stage than in low grade 
and Ta and Tl tumors (p < 0.05) (Table 2). This is consist- 
ent with other findings which reported that the TP53 over- 
expression was associated with high grade and stage of 
bladder cancer [20,46] . 

It has been reported that there is a good concordance 
between overexpression and mutation of TP53 gene 
[20,22], however, other studies have shown a consider- 
able discrepancy between them [47,48]. Analysis of the 
data in this study showed no significant association be- 
tween TP53 mutations and the immunohistochemical 
detection of TP53 protein (p>0.05) (Table 3). This is 
largely because of the four tumors with frameshift muta- 
tions that had no detectable TP53 protein. These muta- 
tions encoded deleted or truncated proteins that are 
very unstable in the cell and usually not detectable by 
IHC method even when using an antibody containing 
the corresponding N-terminal epitope [49,50]. The 
tumors with missense mutations were positive for TP53 
protein overexpression (Table 3); missense mutations 
resulting in amino acid change render the TP53 protein 
a more stable compound with a longer half-life that can 
be detected by standard immunhistochemical methods 
[20,44]. 

On the other hand, the presence of other tumors that 
demonstrated TP53 overexpression without mutations 
might be due to mutations in the TP53 that occurred 
outside exons 5-8 [49], or to the overexpression of 
TP53 caused not only by TP53 mutations but also by 
other factors (such as MDM2) which bind to TP53 pro- 
tein, thus increasing its half-life and allowing it to accu- 
mulate in the nucleus. This observation may reflect 
alterations in the TP53 pathway rather than in the TP53 
gene itself [6,51]. However, the antibody used in our 
study recognises both the wild-type and the mutant 
forms of TP53 protein at the same time. Hence, it is 
possible that the IHC assay detects accumulation of the 
wild-type in some cases. The level of wild-type TP53 
protein can be increased in response to DNA damage, 
hypoxia, oncogene activation and changes in the nucleo- 
tide pool, which are commonly observed in primary 
tumors [51,52]. Furthermore, up-regulation of wild-type 
TP53 protein in tumors may indicate the last step of de- 
fence before metastasis [53]. Therefore, the TP53 alter- 
ation (TP53status) that is used as a prognostic marker of 
bladder tumorigenesis should be determined by combin- 
ation of mutational and IHC analysis. Accordingly, the 



Al-Kashwan et al. BMC Research Notes 2012, 5:466 
http://www.biomedcentral.eom/1756-0500/5/466 



Page 9 of 10 



present investigation of TP53 status has shown a strong 
association with deep invasive tumors (p< 0.01) (Table 2). 
The altered expression of TP53 tumor suppressor gene 
is an independent predictor of bladder cancer progres- 
sion when examined as an individual determinant 
[10,20]. 

Conclusions 

The TP53 frameshift mutations (especially insertion A) 
and 196 hotspot codon may represent a possible specific- 
mutational patterns associated with bladder tumorigen- 
esis, and reflect a preferential target for exogenous carci- 
nogens. Furthermore, the difference in incidence and type 
of TP53 mutations among the Iraqi TCC patients may ex- 
plicitly indicate a distinct molecular pathway responsible 
for the development of bladder cancer due to exposure to 
environmental hazards (e.g. depleted uranium). The un- 
usual mutation patterns of TP53 necessitates a complete 
molecular epidemiological study for further clarification of 
distinct molecular pathways for bladder cancer pathogen- 
esis among Iraqi patients. The current study confirmed 
that the combination of molecular analysis and protein ex- 
pression of TP53 tumor suppressor gene is highly recom- 
mended for studying gene alterations in bladder cancer 
rather than the application of a single approach. 

Abbreviations 

LSAB+: Labeled Streptavidin-biotin; BC: Bladder Cancer; TCC: Transitional Cell 
Carcinoma; FFPE: Formalin-Fixed Paraffin-Embedded Tissue; DU: Depleted 
Uranium; IHC: Immunohistochemistry; PCR: Polymerase chain reaction; 
TUR: Transurethral; SSCP: Single strand conformation polymorphism. 

Competing interests 

The authors declare that they have no competing interests. 
Authors' contributions 

ThAA and MH carried out the molecular genetic studies. THAA performed 
the statistical analysis. AA and AKM initiated the project at the University of 
Kufa . DA and AA carried out the immunohistochemical analysis and 
histopathological examination. MR participated in the molecular genetic 
studies. MNA provided the surgical specimens. AAY and AA conceived of the 
study, participated in its design and drafted the manuscript. All authors read 
and approved the final manuscript. 

Author details 

1 Middle Euphrates unit for cancer research, Faculty of Medicine, University of 
Kufa, Kufa, Iraq. 2 National Institute for Genetic Engineering and 
Biotechnology, Tehran, Iran, department of Pathology and Forensic 
Medicine, Faculty of Medicine, University of Kufa, Kufa, Iraq, biotechnology 
Department, College of Science, Baghdad university, Baghdad, Iraq. 5 Urology 
department, Faculty of Medicine, University of Kufa, Kufa, Iraq, department 
of Pathology and Forensic Medicine, Faculty of Medicine, University of Kufa, 
Kufa, P.O. Box 21, Najaf Governorate, Iraq. 

Received: 3 June 2012 Accepted: 17 August 2012 
Published: 28 August 2012 

References 

1. Bray F, Sanklla R, Ferlay J, Parkin DM: Estimates of cancer incidence and 
mortality in Europe in 1995. Eur J Cancer 2002, 38:99-166. 

2. Heijden A, Hulsbergen-Van de kaa CA, Witjes JA: The influence of thermo- 
chemotherapy on bladder tumors: an immunohistochemical analysis. 

World J Uorl 2007, 25(3):303-308. 



3. Al-Foudi A, Parkin DM: Seven years data from the Baghdad tumor 
registry. Int. J. Cancer 2006, 34(2) doi:1 0.1 002/ijc.291 034021 1. 

4. Zeegers M, Swaen G, Kant I, Goldbohm R, van den Brandt P: Occupational 
risk factors for male bladder cancer: results from a population based 
case cohort study in the Netherlands. Occup Environ Med 2010, 
58:590-596. 

5. Wallerand H, Bakkar AA, de Medina SG, Pairon JC, Yang YC, Vordos D, 
Bittard H, Fauconnet S, Kouyoumdjian JC, Jaurand MC, Zhang ZF, Radvanyi 
F, Thiery JP, Chopin DK: Mutations in TP53, but not FGFR3, in urothelail 
cell carcinoma of the bladder are influenced by smoking: contribution of 
exogenous versus endogenous carcinogens. Carcinogenesis 2005, 
26(1):177-184. 

6. Vogelstein B, Lane D, Levine AJ: Surfing the p53 network. Nature 2000, 
408:307-310. 

7. Olivier M, Eeles R, Hollstein M, Khan MA, Harris CC, Hainaut P: The IARC 
TP53 database: new online analysis and recommendations to users. 
Hum Mutat 2002, 19:607-614. 

8. Chen D, Yu Z, Zhu Z, Lopez C: The p53 pathway promotes efficient 
mitochondrial DNA base excision repair in colorectal cancer cells. 
Cancer Res 2006, 66(7):3485-3494. 

9. Esriq D, Elmajian D, Groshen S, Freeman JA, Stein JP, Chen SC, Nichols PW, 
Skinner DG, Jones PA, Cote RJ: Accumulation of Nuclear p53 and Tumor 
Progression in Bladder Cancer. New Eng J Med 1994, 331 (19):1 259-1 264. 

10. Chatterjee S, Datar R, Youssefzadeh D, George B, Goebell P, Stein J, Young L, 
Shi SR, Gee C, Groshen S, Skinner DG, Cote RJ: Combined effects of P53, 
P21, and PRb expression in the progression of bladder transitional cell 
carcinoma. J Clin Oncol 2004, 22(6):1 007-1 01 3. 

1 1 . Mitra AP, Datar RH, Cote RJ: Molecular staging of bladder cancer. BJU Int 
2005, 96(1):7-12. 

12. Crow JF: How much do you know about spontaneous mutation rates. 

Environ Mol Mutagenesis 1993, 21:122-129. 

13. Hollstein M, Rice K, Greenbelt MS, Soussi T, Fuchs R, Sorlie T, Hoviq E, Smith- 
Sorensen B, Montesano R, Harris CC: Database of p53 gene somatic 
mutations in human tumors and cell lines. Nucleic Acids Res 1994, 
22(17)3351-3355. 

14. Cho Y, Gorina S, Jeffrey PD, Pavletich NP: Crystal structure of a p53 tumor 
suppressor-DNA complex: understanding tumorigenic mutations. 
Science 1994, 265(5170)346-355. 

15. Yamamoto S, Romanenko A, Wei M, Masuda C, Zaparin W, Vinnichenko W, 
Vozianov A, Lee CC, Morimura K, Wanibuchi H, Tada M, Fukushima S: 
Specific p53 gene Mutations in Urinary Bladder Epithelium after the 
Chernobyl Accident. Cancer Res 1999, 59(15)3606-3609. 

16. Vahakangas K: Molecular epidemiology of human cancer risk. Gene- 
enviroment interactions and P53 mutation spectrum in human ling 
cancer. Methods Mol Med 2003, 74:43-59. 

17. Silverman DT, Mooison AS, Deveas SS: Bladder cancer. In Cancer 
epidemiology and prevention. Edited by Schottenfold D, Fraumeni JF. New 
York: Oxford University Press; 1996:1156-1179. 

18. Loda M, Cukor B, Tarn SW, Lavin P, Fiorentino M, Draetta GF, Jessup JM, 
Pagano M: Increased proteasome-dependent degradation of the cyclin- 
dependent kinase inhibitor P27 in aggressive colorectal carcinomas. 
Nature Med 1997, 3(2):231 -234. 

19. Chatterjee SJ, George B, Goebell P, Alavi-Tafreshi M, Shi SR, Fung YK, 
Jones PJ, Cordon-Cardo C, Datar RH, Cote RJ: Hyperphosphorylation of 
pRb: a mechanism for RB tumor suppressor pathway inactivation in 
bladder cancer. J Pathol 2004, 203(3)762-770. 

20. Esriq D, Spruck C 3rd, Nichols PW, Chaiwun B, Steven K, Groshen S, Chen 
SC, Skinner DG, Jones PA, Cote RJ: P53 nuclear protein accumulation 
correlates with mutations in the P53 gene, tumor grade, and stage in 
bladder cancer. Am J Pathol 1993, 143(5):1 389-1 397. 

21. Mizobuchi S, Furihata M, Sonobe H, Ohtsuki Y, Ishikawa T, Murakami H, 
Kurabayashi A, Ogoshi S, Sasaguri S: Association between p53 
immunostaining and cigarette smoking in squamous cell carcinoma 
of the esophagus. Japn J Clin Oncol 2000, 30(10):423-428. 

22. Vet JA, Brinquier PP, Schaafsma HE, Witjes JA, Debruyne FM, Schalken JA: 
Comparison of p53 protein overexpression with p53 mutation in bladder 
cancer: Clinical and biological aspects. Lab Invest 1995, 73(6):837-843. 

23. Xing EP, Yang GY, Wang LD, Shi ST, Yang CS: Loss of heterozygosity of the 
Rb gene correlates with pRb protein expression and associates with p53 
alteration in human esophageal cancer. Clin Cancer Res 1999, 

5(5):1 231 -1240. 



Al-Kashwan et al. BMC Research Notes 201 2, 5:466 Page 1 0 of 1 0 

http://www.biomedcentral.eom/1756-0500/5/466 



24. 



25. 



26. 



27. 



29. 



30. 



31. 



32. 



33. 



Epstein Jl, Amin MB, Reuter VR, Mostofi FK: The World Health 
Organization/International Society of Urological Pathology consensus 
classification of urothelial (transitional cell) neoplasms of the urinary 
bladder. Bladder Consensus Conference Committee. Am J Surg Pathol 
1998, 22(1 2):1 435-1 448. 

Green F, Page D, Fleming I, Fritz A, Balch C, Haller D, Morrow M (Eds): AJCC 
Cancer Staging Manual. 6th edition. New York, NY: Springer; 2002. 
Al-Dujaily EA, Al-Janabi AA, Pierscioniek T, Yasseen AA: High prevalence of 
HER-2/neu overexpression in female breast cancer among an Iraqi 
population exposed to depleted Uranium. Journal of Carcinogenesis 2008, 
7:1-5. 

Al-Abbassi DS, Al-Janabi AA, Al-Toriahi KM, JaborTH, Yasseen AA: 
Expresssion of VEGF in urinary bladder transitional cell carcinoma in an 
Iraqi population subjected to depleted Uranium. Applied 
immunohistochemistry and molecular morphology 2009, 1 7(4):307-31 1 . 
Al-Mumen MM, Al-Janabi AA, Jumaa AS, Al-Toriahi KM, Yasseen AA: 
Exposure to depleted Uranium does nor alter the co-expression of HER- 
2/neu and P53 in breast cancer patients. BMC Research Notes 201 1, 4:87. 
Sato T, Tanigami A, Yamakawa K, Akiyama F, Kasumi F, Sakamoto G, 
Nakamura Y: Allelotype of breast cancer: cumulative allele losses 
promote tumor progression in primary breast cancer. Cancer Res 1990, 
50:7184-7189. 

Liechti-Gallati S, Schneider V, Neeser D, Kraemer: Two buffer page system- 
based SSCP/HD analysis of DNA and RNA: a general protocol for rapid 
and sensitive mutation screening in cystic fibrosis and any other human 
genetic disease. Eur J Hum Genet 1999, 7:590-598. 
Khudair A, Abdul Kader K and Al-Taha T: Study of the radiological pollution 
level in pastures of Basrah in: The conference on the effects of the use of DU 
weaponry on human and environment in Iraq: 26-27March 2002. Iraq: 
Baghdad; 2000. 

Tawfiq NF, Al-Jobori SM, Al-Saji AW, Itawi RK: Determination of Alpha-emitters 
in Iraqi soil samples using solid state nuclear track detectors CR-39 and CN-85. 
The Conference on the Effects of DU Weaponry on Human and Environment in 
Iraq. Baghdad, Iraq: The Iraqi Ministry of Higher Education and Scientific 
Research; 2002. 

Al-Azzawi S: Depleted Uranium radioactive contamination in Iraq: An overview. 
Global Res:. Center for Research on Globalization; 2006. www.globalresearch. 



pathways for bladder cancer in Ukraine, before and after the Chernobyl 
disaster. Oncol Rep 2004, 1 1:881-886. 

46. Shariat SF, Tokunaga H, Zhou J, Kim J, Ayala GE, Benedict WF, Lemer SP: 
P53, P21, PRB, and P16 expression predict clinical outcome in 
cystectomy with bladder cancer. J Clin Oncol 2004, 122(6):1 01 4-1 024. 

47. Matsushima AY, Cesarman E, Chadburn A, Knowles D: Post thymic T cell 
lymphomas frequently overexpress p53 protein but infrequently exhibit 
p53 gene mutation. Am J Pathol 1994, 144(3):573-584. 

48. Hall PA, Lane DP: P53 in tumor pathology: can we trust 
immunohistochemistry? — Revisited. J Pathol 1994, 172(1):1-4. 

49. Greenblatt MS, Bennett WP, Hollstein M, Harris CC: Mutations in p53 tumor 
suppresser gene: clues to cancer etiology and molecular pathogenesis. 
Cancer Res 1994, 54:4855-4878. 

50. Knowles MA: Molecular subtypes of bladder cancer: Jekyll and Hyde or 
chalk and chees? Carcinogenesis 2006, 27(3)361 -373. 

51. Vazquez A, Bond E, Levine A, Bond G: The genetics of the p53 pathway, 
apoptosis and cancer therapy. Nat Rev Drug Discovery 2008, 7:979-987. 

52. Shiao YH, Palli D, Caporaso NE, Alvord WG, Amorosi A, Nesi G, Saieva C, 
Masala G, Fraumeni JF Jr, Rice JM: Genetic and immunohistochemical 
analyses of p53 independently predict regional metastasis of gastric 
cancers. Cancer Epidemiol Biomarkers Prev 2000, 9(6):63 1-633. 

53. Jen K-Y, Cheung VG: Identification of novel p53 target genes in ionizing 
radiation response. Cancer Res 2005, 65:7666-7673. 



doi:1 0.1 1 86/1 756-0500-5-466 

Cite this article as: Al-Kashwan et al:. Specific-mutational patterns of p53 
gene in bladder transitional cell carcinoma among a group of Iraqi 
patients exposed to war environmental hazards. BMC Research Notes 
2012 5:466. 



34. 



35. 
36. 



37. 



39. 



40. 



41. 



42. 



43. 



44. 



45. 



Denissenko MF, Chen JX, Tang M-S, Pfeifer GP: Cytosine methylation 
determines hot spots of DNA damage in the human p53 gene. Proc Natl 
Acad Sci USA 1997, 94:3893-3898. 

Little JB: Radiation Carcinogenesis. Carcinogenesis 2000, 21(3):347-404. 
Lipponen PK: Overexpression of p53 nuclear oncoprotein in transitional 
cell bladder cancer and its prognostic value. Int J Cancer 1993, 
53(3)365-376. 

Yamamoto S, Tatematsu M, Yamamoto M, Fukami H, Fukushima S: Clonal 
analysis of urothelial carcinomas in C3H/HeN <->BALB/c chimeric mice 
treated with N-butyl-N-(4-hydroxybutyl) nitrosamine. Carcinogenesis 1998, 
19(5):855-860. 

Williamson MP, Elder PA, Knowles MA: The spectrum of Tp53 mutations in 
bladder carcinoma. Genes chromosome Cancer 1994, 9(2):1 08-1 1 8. 
Pfeifer GP: P53 mutational spectra and the role of methylated CpG 
sequences. Mutat Res 2000, 450(1 -2):1 55-1 66. 

Schroeder JC, Conway K, Li Y, Mistry K, Bell DA, Taylor JA: p53 Mutations in 
bladder cancer: evidence for exogenous versus endogenous risk factors. 

Cancer Res 2003, 63(21)7530-7538. 

Jung I, Messing E: Molecular mechanism and pathways in bladder cancer 
development and progression. Cancer control 2000, 7(4):325-334. 
Habuchi T, Takahashi R, Yamada H, Ogawa O, Kakehi Y, Ogura K, Hamazaki 
S, Toguchida J, Ishizaki K, Fujita J: Influence of cigarette smoking and 
schistosomiasis on p53 gene mutation in urothelial cancer. Cancer Res 
1993,53(16)3795-3799. 

Harris CC: Structure and function of the p53 tumor suppressor gene: 
clues for rational cancer therapeutic strategies. J Natl Cancer Inst 1996, 
88(20):1 442-1 455. 

Moore LE, Smith AH, Eng C, DeVries SD, Kalman D, Bhargava V, Chew S, 
Ferreccio C, Rey OA, Hopenhayn C, Biggs ML, Bates MN, Waldman FM: P53 
alterations in bladder tumors from arsenic and tobacco exposed 
patients. Carcinogenesis 2003, 24(1 1):1 785-1 791. 
Morimura K, Romanenko A, Min W, Salim E, Kinoshita A, Wanibuchi H, 
Vozianov A, Fukushima S: Possible disitinct molecular carcinogenic 



Submit your next manuscript to BioMed Central 
and take full advantage of: 

• Convenient online submission 

• Thorough peer review 

• No space constraints or color figure charges 

• Immediate publication on acceptance 

• Inclusion in PubMed, CAS, Scopus and Google Scholar 

• Research which is freely available for redistribution 



Submit your manuscript at 
www.biomedcentral.com/submit 



o 



BioMed Central